Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Biochem Mol Toxicol ; 37(5): e23318, 2023 May.
Article in English | MEDLINE | ID: mdl-36762617

ABSTRACT

This work aimed to investigate the role and mechanism of NADPH oxidase 4 (NOX4) in the polarization of microglial cells. Microglial cells were transfected with the NOX4 overexpression plasmid (pGL3-NOX4), and later treated with lipopolysaccharide (LPS) and interferon-γ (IFN-γ) to induce its M1 polarization. Later, the F4/80 + CD86 + cell proportion was detected by flow cytometry (FCM), the inflammatory factor expression levels were analyzed through enzyme-linked immunosorbent assay (ELISA), while ionized calcium binding adapter molecule 1 (IBA-1) and PKM2 expression were measured by immunofluorescence (IF) staining. In addition, dichlorodihydrofluorescein diacetate probe was utilized to detect the reactive oxygen species (ROS) levels, glucose uptake, and glycolysis, as well as lactic acid level. The expression of glycolytic enzymes PKM2, HK2, and citrate (Si)-synthas (CS) was detected by Western-blot (WB) assay. Moreover, the polarization level of microglial cells was detected after ROS expression was suppressed by the ROS inhibitor N-acetylcysteine (NAC). In mouse experiments, LPS was applied in inducing central neuroinflammation in NOX4 knockdown mouse model (KO) and wild-type mice (WT). Thereafter, the inflammatory factor levels and lactic acid level in mouse tissues were detected; IBA-1 and CD86 expression in mice was measured by IF staining; and the expression of glycolytic enzymes PKM2, HK2, and CS in the central nervous system (CNS) was also detected. After NOX4 overexpression in microglial cells, the M1 polarization level was upregulated, the F4/80 + CD86 + cell proportion increased, and inflammatory factors were upregulated. At the same time, the expression of glycolytic enzymes PKM2, HK2, and CS was upregulated. NAC pretreatment suppressed the effects of NOX4, reduced the F4/80 + CD86 + cell proportion, and suppressed the expression of PKM2, HK2, and CS. In the mouse model, the expression levels of CD86 in KO group decreased, and the inflammatory factors were also downregulated. NOX4 promotes glycolysis of microglial cells via ROS, thus accelerating M1 polarization and inflammatory factor expression. In this regard, NOX4 is promising as a new target for the treatment of neuroinflammation.


Subject(s)
Glycolysis , Microglia , NADPH Oxidase 4 , Neuroinflammatory Diseases , Animals , Mice , Lipopolysaccharides , Microglia/metabolism , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , Reactive Oxygen Species/metabolism
2.
J Cell Mol Med ; 26(9): 2706-2716, 2022 05.
Article in English | MEDLINE | ID: mdl-35352469

ABSTRACT

Antrodia Camphorata Polysaccharide (ACP) refers to a kind of polysaccharide extracted from the natural porous fungus Antrodia camphorata. This study investigated the mechanism of action of ACP in protecting the liver. The results showed that ACP suppressed the LPS-induced KC cell activation, reduced the expression of inflammatory factors, increased the SOD level and suppressed ROS expression. In addition, N-acetylcysteine (NAC) was adopted for pre-treatment to suppress ROS. The results indicated that NAC synergistically exerted its effect with ACP, suggesting that ACP played its role through suppressing ROS. Further detection revealed that ACP activated the Nrf2 signal. It was discovered in the mouse model that, ACP effectively improved liver injury in mice, decreased ALT and AST levels, and suppressed the expression of inflammatory factors. This study suggests that ACP can exert its effect against oxidative stress via the Nrf2-ARE signalling, which further improves the production of ROS and the activation of TLR4-NF-κB signalling, and protects the liver against liver injury.


Subject(s)
Antrodia , NF-kappa B , Animals , Antrodia/metabolism , Liver/metabolism , Mice , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Polyporales , Polysaccharides/metabolism , Polysaccharides/pharmacology , Reactive Oxygen Species/metabolism , Toll-Like Receptor 4/metabolism
3.
J Biochem Mol Toxicol ; 36(11): e23188, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35924425

ABSTRACT

This study focuses on exploring the role and mechanism of moronic acid (MOA), a small triterpenoid molecule, against inflammatory bowel disease (IBD). Intestinal macrophages were cultured in vitro, and their M1 polarization was induced by lipopolysaccharide (LPS) and interferon gamma (IFN-γ). After intervention with MOA, the proportion of M1 macrophages was detected, and the levels of inflammatory cytokines (TNF-α, IL-6, and IL-1ß) were examined by ELISA. IFA staining was performed to determine the P50 and CD86 expressions, while DCFH-DA was used to determine the reactive oxygen species (ROS) level, as well as the p-P50 and NLRP3 protein levels. Additionally, we also used N-acetylcysteine, a ROS inhibitor, to further explore the association between MOA and ROS-NF-κB signaling. In murine experimentation, colitis was induced in mice with DSS. After MOA intervention, we assessed the mucosal barrier damage, tissue ROS, as well as protein and inflammatory cytokine levels. MOA could inhibit the M1 polarization of intestinal macrophages, suppress the expressions of inflammatory cytokines, and reduce the level of ROS-NF-κB-NLRP3 signaling. After inhibiting ROS through NAC treatment, the effect of MOA was evidently weakened. Clearly, MOA exerted its activity via ROS. In the murine model, MOA could lower the CD86 level in the intestinal tissues, inhibit the M1 polarization of macrophages, and reduce the tissue levels of inflammatory cytokines. This study finds that MOA can regulate ROS-NF-κB-NLRP3 signaling by inhibiting ROS, thereby suppressing the M1 polarization of intestinal macrophages, which plays a protective role in IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Mice , Animals , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein , Reactive Oxygen Species , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Cytokines/metabolism , Macrophages/metabolism , Lipopolysaccharides/toxicity , Inflammation/drug therapy
4.
J Cell Mol Med ; 25(20): 9513-9522, 2021 10.
Article in English | MEDLINE | ID: mdl-34464498

ABSTRACT

Piperine (PIP), the main active ingredient in pepper, belongs to the cinnamamide alkaloid. PIP has been found to have functions, including anti-oxidation, immune regulation, anti-tumour and promotion of drug metabolism. The present study was mainly designed to reveal the anti-tumour effect of PIP against gastric cancer and the relevant mechanism. In brief, the undifferentiated human gastric cancer cell HGC-27 was used, which was treated with different concentrations of PIP. As a result, PIP could inhibit proliferation and induce apoptosis of HGC-27 cells in a dose-dependent manner. The mechanism of PIP was associated with ROS increase and mitochondrial damage, simultaneously, the expression of key proteins of apoptosis was affected, including Bcl-2, Bax, Cyt-c, Caspase-9 and Caspase-3. Pre-treatment of ROS scavenger NAC HGC-27 cells could significantly reduce PIP-induced apoptosis and inhibit the activation of apoptotic signals. Consistently, PIP could induce ROS to increase and activate apoptotic signals in the animal model. Therefore, the present study showed that PIP can induce the generation of ROS, thereby promoting the activation of mitochondrial apoptotic pathway and exerting anti-tumour effects.


Subject(s)
Alkaloids/pharmacology , Apoptosis/drug effects , Benzodioxoles/pharmacology , Mitochondria/drug effects , Mitochondria/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Stomach Neoplasms/etiology , Stomach Neoplasms/metabolism , Animals , Biomarkers , Caspase 3/genetics , Caspase 3/metabolism , Caspase 9/genetics , Caspase 9/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Humans , Immunophenotyping , Membrane Potential, Mitochondrial/drug effects , Mice , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays
5.
J Cell Mol Med ; 25(15): 7257-7269, 2021 08.
Article in English | MEDLINE | ID: mdl-34184406

ABSTRACT

Our previous study has found that aureusidin can inhibit inflammation by targeting myeloid differentiation 2 (MD2) protein. Structural optimization of aureusidin gave rise to a derivative named CNQX. LPS was used to induce inflammation in intestinal macrophages; flow cytometry, PI staining and Hoechst 33342 staining were used to detect the apoptotic level of macrophages; enzyme-linked immunosorbent assay (ELISA) was utilized to detect the expression level of inflammatory factors (including IL-1ß, IL-18 and TNF-α); immunofluorescence staining was used to investigate the expression of MD2; Western blot was employed to measure the protein level of TLR4, MD2, MyD88 and p-P65. As a result, CNQX with IC50 of 2.5 µM can significantly inhibit the inflammatory damage of macrophages, decrease apoptotic level, reduce the expression level of inflammatory factors and simultaneously decrease the expression level of TLR4, MD2, MyD88 as well as p-P65. Caco-2 cell line was used to simulate the intestinal mucosal barrier in vitro, LPS was employed to induce cell injury in Caco-2 (to up-regulate barrier permeability), and CNQX with IC50 of 2.5 µl was used for intervention. Flow cytometry was used to detect the apoptotic level of Caco-2 cells, trans-epithelial electric resistance (TEER) was measured, FITC-D was used to detect the permeability of the intestinal mucosa, and Western blot was used to detect the expression levels of tight junction proteins (including occludin, claudin-1, MyD88, TLR4 and MD2). As a result, CNQX decreased the apoptotic level of Caco-2 cells, increased TEER value, decreased the expression levels of MyD88, TLR4 and MD2, and increased the protein levels of tight junction proteins (including occludin and claudin-1). C57BL/6 wild-type mice were treated with drinking water containing Dextran sulphate sodium (DSS) to establish murine chronic colitis model. After CQNX intervention, we detected the bodyweight, DAI score and H&E tissue staining to evaluate the life status and pathological changes. Immunohistochemistry (IHC) staining was used to detect the expression of MD2 protein, tight junction protein (including occludin and claudin-1). Transmission electron microscopy and FITC-D were used to detect intestinal mucosal permeability. Western blot was used to detect the expression levels of tight junction proteins (including occludin, claudin-1, MyD88, TLR4 and MD2) in the intestinal mucosa tissue. Consequently, CNQX can inhibit the intestinal inflammatory response in mice with colitis, inhibit the mucosal barrier injury, increase the expression of tight junction proteins (including occludin and claudin-1) and decrease the expression levels of MyD88, TLR4 and MD2. Mechanistically, pull-down and immunoprecipitation assays showed that CNQX can inhibit the activation of TLR4/MD2-NF-κB by binding to MD2 protein. Collectively, in this study, we found that CNQX can suppress the activation of TLR4 signals by targeting MD2 protein, thereby inhibiting inflammation and mucosal barrier damage of chronic colitis.


Subject(s)
6-Cyano-7-nitroquinoxaline-2,3-dione/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Colitis, Ulcerative/drug therapy , Intestinal Mucosa/drug effects , Lymphocyte Antigen 96/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Caco-2 Cells , Colitis, Ulcerative/metabolism , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Toll-Like Receptor 4/metabolism , Transcription Factor RelA/metabolism
6.
J Cell Physiol ; 234(11): 21089-21099, 2019 11.
Article in English | MEDLINE | ID: mdl-31032920

ABSTRACT

The present study was designed to investigate the mechanism of myeloid differentiation protein 2 (MD2) on intestinal mucosa destruction in mice with chronic colitis. Briefly, a chronic colitis mouse model was established by the administration of dextran sulfate sodium (DSS) in transgenic mice of MD2 overexpression (Transgenic, MD2-Tg) and C57BL/6 wild-type mice (MD2-WT). In addition, Caco-2 cells were cultured to form a monolayer cell model in vitro. The small interfering RNA was utilized to silence the MD2 gene in Caco-2 cells, and tumor necrosis factor-α (TNF-α) was used to establish the model of intestinal mucosal inflammation. After DSS induction, the intestinal mucosal tissue inflammation was more severe in MD2-Tg mice than MD2-WT. In addition, the intestinal mucosa was severely damaged, the intestinal mucosal permeability was increased, bacterial translocation was obvious, and the expression levels of MD2, MyD88, Toll-like receptor 4 (TLR4), and HMGB1 in mucosal tissues were significantly increased, while the expression levels of tight junction proteins, occludin, and claudin-1 were significantly lower in MD2-Tg mice compared with those in MD2-WT mice. TNF-α could induce inflammatory apoptosis in Caco-2 cell models. After MD2 silencing, the apoptotic level was decreased, the value of transepithelial electrical resistance was increased, the permeability of intestinal mucosa was decreased, the cellular expression levels of MD2, MyD88, TLR4, and HMGB1 were decreased, while the expression levels of tight junction proteins, occludin and claudin-1 were increased. MD2 could aggravate the destruction of intestinal mucosa in chronic colitis through the HMGB1-TLR4-MyD88 pathway.


Subject(s)
Colitis/metabolism , Colitis/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lymphocyte Antigen 96/metabolism , Adult , Aged , Animals , Caco-2 Cells , Female , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Permeability , Tight Junctions/metabolism , Tight Junctions/pathology
7.
Neurosurg Rev ; 42(3): 715-720, 2019 Sep.
Article in English | MEDLINE | ID: mdl-30607616

ABSTRACT

The study was designed to investigate the predictive value of phosphorylated CAMP response element binding protein (p-CREB) level in peripheral blood on secondary cognitive impairment in patients with mild-to-moderate craniocerebral trauma. A total of 107 patients with mild-to-moderate craniocerebral trauma were selected, who were admitted to the Second Affiliated Hospital of College of Jiaxing from January 2016 to January 2017. Of them, 30 patients were diagnosed with secondary mild cognitive impairment (MCI) during follow-up, who were assigned to the experimental group. The remaining 77 subjects were assigned to the control group, without significant cognitive impairment. The clinical data of patients were compared between two groups, and the clinical data of patients with different p-CREB levels were compared. Logistic regression analysis was used to investigate the risks of MCI in patients with different p-CREB levels. Moreover, multiple linear regression analysis was employed to assess the influencing factors of scores of Mini-Mental State Examination (MMSE) on patients with secondary MCI. The following pathophysiologic factors, including age, rescuing time, the proportion of hypertension, trauma severity score (AIS-ISS), and serum total cholesterol (TC) were significantly higher in patients in the experimental group compared to those in the control group (all P < 0.05). The serum level of p-CREB ranged from 0.127 to 1.852 ng/ml. Afterwards, the serum levels of p-CREB of patients were divided into four quartiles. The first, second, third, and fourth quartile groups were 0.127-0.548 ng/ml, 0.549-0.982 ng/ml, 0.983-1.412 ng/ml, and 1.413-1.852 ng/ml, respectively. As the level of p-CREB increased, age, rescuing time, the proportion of hypertension, and AIS-ISS gradually decreased, with statistical significance (all P < 0.05). Univariate and multivariate logistic regression analyses demonstrated that the risk of secondary MCI of patients in the first quartile was 1.21 and 1.58 times of the fourth quarter, respectively. Multivariate linear regression analysis showed that age, rescuing time, AIS-ISS, and serum p-CREB level were independent influencing factors of MMSE score in secondary MCI patients. For each increase of 0.1 ng/ml in serum p-CREB level, the MMSE score increased by 0.382 in MCI patients. Serum p-CREB level was an independent risk factor of secondary MCI in patients with mild-to-moderate craniocerebral trauma, whose level was significantly correlated with the injured degree of cognitive impairment. The level of p-CREB is also age-related, and younger patients have a higher level.


Subject(s)
Cognitive Dysfunction/blood , Cognitive Dysfunction/etiology , Craniocerebral Trauma/complications , Cyclic AMP Response Element-Binding Protein/blood , Adult , Aged , Case-Control Studies , Cognitive Dysfunction/diagnosis , Craniocerebral Trauma/blood , Female , Humans , Hypertension/blood , Hypertension/complications , Male , Middle Aged , Predictive Value of Tests , Risk Factors
8.
Aging (Albany NY) ; 15(14): 6721-6735, 2023 04 04.
Article in English | MEDLINE | ID: mdl-37494667

ABSTRACT

This work aimed to investigate the role of helper T cell 1 (Th1) in chronic colitis and its immunoregulatory mechanism. The proportions of Th1 and Th2, and the levels of related cytokines in tissues from patients with inflammatory bowel disease (IBD; ulcerative colitis+Crohn's disease, UC+CD) were detected. DSS was used to induce the mouse model of IBD; thereafter, Th1 cells were induced in vitro and amplified before they were injected intraperitoneally. Later, the changes in life state and body weight of mice were observed, the proportion of M1 macrophages in mucosal tissues and mucosal barrier damage were detected. After treatment with macrophage scavenging agent (Clodronate Liposomes, CLL), the influence of Th1 on IBD mice was observed. Then, the intestinal macrophages were co-cultured with Th1 in vitro to observe the influence of Th1 on the polarization of intestinal macrophages. Besides, cells were treated with the STAT3 inhibitor to further detect the macrophage polarization level. Intestinal macrophages were later co-cultured with intestinal epithelial cells to observe the degree of epithelial cell injury. The Th1 proportions in intestinal tissues of UC and CD patients were higher than those in healthy subjects, but the difference in Th2 proportion was not significant. In the IBD mouse model, Th1 induced the M1 polarization of macrophages, aggravated the intestinal inflammatory response, and resulted in the increased mucosal barrier permeability. Pretreatment with CLL antagonized the effect of Th1 cells, reduced the intestinal tissue inflammatory response and mucosal barrier permeability.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Leukemia, Lymphocytic, Chronic, B-Cell , Animals , Mice , Intestinal Mucosa , Colitis/chemically induced , Macrophages , Disease Models, Animal
9.
Int Immunopharmacol ; 117: 109992, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37012876

ABSTRACT

This work aimed to investigate the role of long non-coding RNA (lncRNA) PCSK6-AS1 in inflammatory bowel disease (IBD). The levels of PCSK6-AS1 in human samples were detected, and its target protein HIPK2 was explored by protein mass spectrometry and ground select test (GST) method. Meanwhile, the HIPK2-STAT1 interaction relation was verified by pull-down assay. In the mouse model, Dextran Sulfate Sodium(DSS) was used to induce mouse colitis, then the effect of PCSK6-AS1 on mouse mucosal barrier was detected by immunohistochemical (IHC) staining, hematoxylin and eosin (H&E) staining, and the proportion of T-helper cells 1(Th1) cells was measured by flow cytometry (FCM). For in-vitro experiments, Th0 cells were used as the objects, and the effect of PCSK6-AS1 on Th1 differentiation was explored by FCM and enzyme-linked immunosorbent assay (ELISA). According to our results, the expression of PCSK6-AS1 in colitis tissues increased. PCSK6-AS1 interacted with HIPK2 to promote the expression of the latter, while HIPK2 promoted STAT1 phosphorylation to regulate Th1 differentiation. Th1 differentiation accelerated the mucosal barrier injury and aggravated the progression of colitis. In the Th0 model, PCSK6-AS1 promoted Th1 differentiation. In the animal model, PCSK6-AS1 enhanced Th1 differentiation in the tissues, decreased the tight junction (TJ) protein levels, and improved the mucosal barrier permeability. Suppressing PCSK6-AS1 and the HIPK2 inhibitor tBID decreased Th1 differentiation and tissue inflammation. According to our results, PCSK6-AS1 promotes Th1 cell differentiation via the HIPK2-STAT1 signaling, thus aggravating the chronic colitis-related mucosal barrier damage and tissue inflammation. PCSK6-AS1 has an important role in the occurrence and development of IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , RNA, Long Noncoding , Humans , Mice , Animals , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Phosphorylation , Colitis/metabolism , Inflammatory Bowel Diseases/metabolism , Inflammation/metabolism , Cell Differentiation , Intestinal Mucosa , Disease Models, Animal , Dextran Sulfate/pharmacology , STAT1 Transcription Factor/metabolism , Carrier Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism
10.
Aging (Albany NY) ; 14(22): 8970-8981, 2022 10 10.
Article in English | MEDLINE | ID: mdl-36227135

ABSTRACT

This study illustrated the liver protection mechanism of ACP from the perspective of autophagy activation. ACP suppressed the inflammatory injury of KCs, and decreased the cell apoptosis rate. After LTG and LC3 staining, ACP promoted lysosomal production, increased LC3 expression, activated autophagy, and suppressed the expression of NLRP3 and inflammatory factors. Under the electron microscope, ACP accelerated the production of autophagosomes. After simultaneous treatment with 3-MA and ACP, the effect of ACP on resisting KC injury decreased, the expression of NLRP3 was up-regulated, and autophagy was suppressed. As discovered in the mouse model of liver injury, ACP inhibited the ALT and AST levels, promoted the occurrence of autophagy, reduced NLRP3 expression and alleviated liver injury. ACP activates autophagy to induce NLRP3 degradation, thus suppressing inflammatory response in liver injury and exerting the liver protection effect, which is one of the mechanisms of action of ACP.


Subject(s)
Liver , NLR Family, Pyrin Domain-Containing 3 Protein , Mice , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Liver/metabolism , Autophagy , Polysaccharides/pharmacology
11.
Arch Med Sci ; 18(1): 71-78, 2022.
Article in English | MEDLINE | ID: mdl-35154527

ABSTRACT

INTRODUCTION: The study was designed to assess the expression of long non-coding RNA HOTAIR (lncRNA HOTAIR) in tissues and peripheral blood of patients with advanced hepatocellular carcinoma (HCC). In addition, we also investigated the prognostic correlation between the expression level of lncRNA HOTAIR in tumour tissues and peripheral blood of patients with advanced HCC and sunitinib monotherapy. MATERIAL AND METHODS: A total of 60 patients with advanced HCC who received sunitinib monotherapy and another 60 healthy individuals who were examined at the physical examination centre during the same period were included in the study. Real-time quantitative PCR (RT-QPCR) was used to determine the relative expression of lncRNA HOTAIR in tumour tissue, adjacent tissue, and peripheral blood of HCC patients as well as peripheral blood of healthy controls. Moreover, the clinicopathological information, overall survival (OS), and progression-free survival (PFS) were collected, followed by correlation analysis with lncRNA HOTAIR expression. RESULTS: The expression of lncRNA HOTAIR was significantly higher in tumour tissues compared to that in adjacent tissues (t = 9.03, p < 0.001). The expression of lncRNA HOTAIR in peripheral blood of HCC patients was higher than that in healthy controls (t = 8.04, p < 0.001). There was a correlation between the expression of lncRNA HOTAIR in tumour tissue and peripheral blood in HCC patients (r = 0.638, p < 0.001). Patients with low lncRNA HOTAIR expression in tumour tissues harboured significantly longer OS (13.4 vs. 9.5, p < 0.001) and PFS (8.4 vs. 6.2, p < 0.001) compared to those with high expression. Consistently, patients with low lncRNA HOTAIR expression in peripheral blood had significantly prolonged OS (12.8 vs. 9.1, p < 0.001) and PFS (8.9 vs. 6.4, p < 0.001) compared to those with high expression. Patients with low expression both in tumour tissue and peripheral blood had prolonged OS (14.3 vs. 8.8, p < 0.001) and PFS (10.6 vs. 6.0, p < 0.001) compared to the rest of the patients. Cox regression analysis indicated that the expression level of lncRNA HOTAIR in tumour tissue and peripheral blood was an independent predictive factor of OS and PFS in patients with advanced HCC treated by sunitinib. CONCLUSIONS: The expression of lncRNA HOTAIR was up-regulated in tumour tissue and peripheral blood in patients with advanced HCC. In addition, the expression level of lncRNA HOTAIR was one of the indicators predicting the effectiveness of sunitinib therapy.

12.
Aging (Albany NY) ; 14(17): 6905-6916, 2022 Jul 13.
Article in English | MEDLINE | ID: mdl-35832027

ABSTRACT

AIM: This work aimed to investigate the mechanism of NOX4 in promoting Kupffer cells (KCs) activation and tissue inflammatory response in acute liver injury. METHODS: Initially, the mouse KCs were cultured in vitro. Thereafter, the NOX4 overexpression plasmid was transfected into KCs to construct the overexpression cell line. Then, KCs inflammatory response was induced by LPS + Nigericin treatment. CCK-8 assay was performed to detect cell viability, flow cytometry (FCM) was conducted to measure cell apoptosis, enzyme-linked immunosorbent assay (ELISA) was performed to detect inflammatory factor levels in the culture medium, NLRP3 and ASC expression in cells was detected by immunofluorescence (IF) staining, and ROS expression was detected by the DCFH-DA probe. Furthermore, the expression levels of NLRP3, ASC and Caspase-1 proteins were detected by Western-Blot (WB) assay. Furthermore, cells were pre-treated with NOX inhibitor or NAC to suppress NOX4 expression or ROS production, aiming to further investigate the effect on KCs inflammatory response. In mouse experiments, the NOX4 knockdown mice and wild-type (WT) mice were adopted for carrying out experiments. The mouse model of ALI was constructed with LPS and D-GalN treatment. Thereafter, the changes in tissue samples were detected by H&E staining, NLRP3 expression was measured by histochemical staining, inflammatory factors in tissues were analyzed by ELISA, and the levels of NLRP3, ASC and Caspase-1 proteins in tissues were detected by WB assay. RESULTS: LPS induced KCs inflammatory response. NOX4 overexpression decreased the mouse viability and increased the apoptosis rate. The levels of inflammatory factors were up-regulated in the culture medium. In addition, ROS were activated, and the positive cell number increased. Moreover, NOX4 promoted NLRP3 activation and significantly increased the expression of NLRP3 and ASC. Pretreatment with NOX4 inhibitor or NAC antagonized the effects of NOX4 and suppressed the KCs inflammatory response. In the mouse model, NOX4 knockdown significantly suppressed the activation and inflammatory response of microglial cells in tissues, reducing the NLRP3 expression in tissues. CONCLUSION: NOX4 activates the NLRP3 inflammasome via ROS to promote inflammatory response in KCs and the release of inflammatory factors, suppressing NOX4 can improve ALI in mice, and NOX4 is promising as a new target for ALI treatment.


Subject(s)
Inflammasomes , Kupffer Cells , Animals , Caspase 1/metabolism , Disease Models, Animal , Inflammasomes/metabolism , Kupffer Cells/metabolism , Lipopolysaccharides/pharmacology , Liver/metabolism , Mice , NADPH Oxidase 4/genetics , NADPH Oxidase 4/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nigericin/metabolism , Nigericin/pharmacology , Reactive Oxygen Species/metabolism
13.
Aging (Albany NY) ; 13(17): 21470-21482, 2021 09 07.
Article in English | MEDLINE | ID: mdl-34491906

ABSTRACT

Short-chain fatty acids (SCFAs) are a product of intestinal bacteria metabolism. Our previous study has found that intestinal bacteria in patients with Alzheimer's disease (AD) can promote the activation of NLRP3 inflammasome and mediate neuroinflammation. In this study, we mainly explored the regulation of intestinal microenvironmental immunity by intestinal bacterial metabolite SCFAs and the mechanism of NLRP3 activation. First, wild-type (WT) and APP/PS1 mice were intervened with SCFAs. As a result, the proportion of double-negative T cells (CD3+CD4-CD8-, DNTs) in the intestine was increased, SCFAs could promote the expression of intestinal NLRP3 and inflammatory factors (IL-18, IL-6 and TNF-α). Moreover, SCAFs could also promote the level of inflammatory factors in the cerebrospinal fluid (CSF) of mice and aggravate the cognitive impairment in AD mice. CD3+ T cells isolated from the spleen were pre-treated with SCFAs, followed by detection of the proportion of DNTs. Consequently, SCFAs could promote the formation of DNTs, activate OX40 signal and simultaneously up-regulate the protein expression of Bcl-2, Bcl-xl and Survivin. Knockdown of OX40 could inhibit SCFAs-induced differentiation of DNTs. The co-culture of DNTs and intestinal macrophages showed that DNTs could activate Fas/FasL-TNF-α signal and induce the activation of NLRP3 inflammasome. In AD mouse models, treatment with Fas and TNFR1 inhibitors could significantly inhibit SCFAs-induced NLRP3 activation and inflammatory factors, while attenuate the inflammatory response in the brain tissue of mice and improve the cognitive ability of mice, however, without significant effect on the level of DNTs. The present study showed that SCFAs can promote the formation of DNTs through OX40. DNTs could induce the activation of NLRP3 inflammasome and the release of inflammatory factors in macrophages through Fas/FasL-TNF-α signals, thereby increasing the level of inflammatory factors in the central nervous system. When Fas and TNFR1 were inhibited by suppressing the functions of DNTs and macrophages, the activation of NLRP3 was inhibited. DNTs are affected by SCFAs, which is a new mechanism of neuroinflammation in AD.


Subject(s)
Fatty Acids, Volatile/metabolism , Inflammasomes/metabolism , Intestines , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Neuroinflammatory Diseases/metabolism , T-Lymphocytes/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Brain/metabolism , Brain/pathology , Cytokines/metabolism , Disease Models, Animal , Fas Ligand Protein/metabolism , Inflammation , Intestines/immunology , Intestines/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Receptors, OX40/metabolism , Tumor Necrosis Factor-alpha/metabolism , fas Receptor/metabolism
14.
Int Immunopharmacol ; 101(Pt B): 108290, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34717194

ABSTRACT

Oridonin (Ori) is a kind of diterpenoid small molecule, but its role in nonalcoholic fatty liver disease (NAFLD) has not been reported yet. This study aimed to explore the pharmacological function of Ori in liver protection through the reactive oxygen species (ROS)-mediated polarization of Kupffer cells (KCs). In the present work, KCs were adopted for study in vitro. To be specific, LPS and IFN-γ were utilized to induce M1 polarization, then the influence of Ori intervention on the expression of inflammatory factors IL-1ß, IL-6 and TNF-α was detected by enzyme-linked immunosorbent assay (ELISA), that of CD86 and P65 was measured through fluorescence staining, that of p-P65 and p-P50 was detected by Western blotting (WB) assay, and ROS expression was measured by using the DCFH-DA probe. The C57BL/6J mice were fed with the high fat diet (HFD) to construct the NAFLD model, and intervened with Ori. The blood glucose (BG), body weight (BW), food intake and water intake of mice were monitored; meanwhile, glucose and insulin tolerance tests were conducted. The liver tissues of mice were subjected to H&E staining and oil red O staining. Moreover, the serum ALT, AST and TG levels in mice were monitored, the CD86 and CD206 levels were measured through histochemical staining, the expression of inflammatory factors was detected by ELISA, and the p-P65 and p-P50 protein levels were detected by WB assay. Ori suppressed the M1 polarization of KCs, reduced the levels of inflammatory factors, and decreased the expression of ROS, p-P65 and p-P50. In animal experiments, Ori improved lipid deposition and liver injury in the liver tissues of NAFLD mice, increased the proportion of M2 cells (up-regulated CD206 expression), reduced that of M1 cells (down-regulated CD86 expression), and decreased the serum ALT, AST and TG levels. This study discovered that Ori suppressed ROS production and regulated the M1 polarization of KCs, thus protecting the liver in NAFLD.


Subject(s)
Non-alcoholic Fatty Liver Disease/metabolism , Animals , Diet, High-Fat , Diterpenes, Kaurane , Kupffer Cells/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Protective Agents , Reactive Oxygen Species/metabolism , Signal Transduction , Transcription Factor RelA
15.
Int Immunopharmacol ; 100: 108126, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34492534

ABSTRACT

Pyroptosis, a pattern of inflammatory death, is regulated by NLRP3-Caspase-1 inflammasome and GSDMD-FL protein. Antcin A is a small triterpenoid molecule. In this study, Kupffer cells (KC) were used for in vitro model, which were treated with LPS and Nigericin (L/N) to induce pyroptosis. ELISA was used to determine the influence of Antcin A on the expression of inflammatory factors, IF was utilized to investigate NLRP3 and Caspase-1, PI staining was used to detect the opening level of membrane pores in KCs, C57BL/6J wild-type mice were fed with high-fat diet to construct a NAFLD model, and were simultaneously treated with Antcin A. H&E staining was used to detect hepatic pathological changes in mice, oil red staining was utilized to detect hepatic fat deposits in mice, IHC was used to detect the expression of NLRP3 and Caspase-1, Western blot was used to detect the expression levels of NLRP3 inflammasome (including NLRP3, ASC, Caspase-1, GSDMD-FL and GSDMD-NT). Pull-down assay and immunoprecipitation assay were used to detect the binding between Antcin A and NLRP3. As a result, Antcin A could significantly inhibit the occurrence of pyrolysis, decrease the expression of inflammatory factors, inhibit the activation and assembly of NLRP3 inflammasome, and significantly down-regulate the expression of NLRP3, Caspase-1 and GSDMD-NT in KCs. In NAFLD mice, Antcin A could suppress the inflammatory response in liver tissues of mice, reduce lipid deposition, down-regulate the levels of ALT and AST, and improve liver function in mice. Antcin A could also inhibit the activation of NLRP3 inflammasome in liver tissue and decrease the level of inflammatory factors. In the study of mechanism, we revealed that Antcin A could inhibit the assembly and activation of NLRP3 inflammasome by binding with NLRP3. In summary, in this study, we found that Antcin A could inhibit pyroptosis in KC and alleviate the inflammatory response of liver tissue in NAFLD by targeting NLRP3 inflammasome, which was one of the mechanisms of Anctin A in protecting liver.


Subject(s)
Kupffer Cells/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Pyroptosis/drug effects , Steroids/pharmacology , Animals , Caspase 1/metabolism , Cell Survival , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Inflammasomes/metabolism , Inflammation , Interleukin-1beta/metabolism , Lipopolysaccharides , Liver/pathology , Mice , Mice, Inbred C57BL , Nigericin , Non-alcoholic Fatty Liver Disease/pathology
16.
Front Pharmacol ; 12: 752118, 2021.
Article in English | MEDLINE | ID: mdl-34955826

ABSTRACT

Background: In this study, we mainly aimed to explore the correlation between galloflavin and NLRP3 and its effect on colorectal cancer. Methods: NLRP3 was overexpressed in SW480 cells; LPS + ATP was used to mimic the inflammatory microenvironment. Wound healing assay and Transwell assay were utilized to detect cell migration and invasion abilities; CCK-8 assay was performed to detect cell viability alterations; colony formation assay was conducted to detect colony formation ability; Western blot was used to detect the levels of NLRP3, ASC, C-Myc, and P21. SW480 cells were pretreated with high-dose and low-dose galloflavin, followed by observation of their effects on cell metastasis and invasion. NLRP3 was knocked out in SW480 to construct the SW480-NLRP3-/- cell line, followed by high-dose galloflavin treatment and subsequent observation of cell metastasis and invasion abilities. Small molecule-protein docking and pull-down assay were performed to confirm the targeting relationship between galloflavin and NLRP3. After constructing a tumor-bearing mice model, galloflavin was intragastrically administered, followed by detection of tumor growth, expression of NLRP3 and ASC by immunohistochemistry, and tumor histopathology by H&E staining. Results: After NLRP3 overexpression and LPS/ATP induction in SW480, the cell migration and invasion abilities were significantly enhanced, and cell viability was also enhanced. The activation of NLRP3 could promote the malignant behavior of colorectal cancer cells in the inflammatory microenvironment. Galloflavin treatment could significantly attenuate the malignant behavior of SW480 in the inflammatory microenvironment and inhibit the migration and invasion capabilities of SW480. The knockout of NLRP3 inhibited the effect of galloflavin, which did not significantly change the migration and invasion abilities. Molecular docking and pull-down assay revealed a targeted binding relationship between galloflavin and NLRP3 and that galloflavin is bound to NLRP3 not ASC protein. Moreover, galloflavin could inhibit tumor growth and decrease the expression of NLRP in tumor-bearing mice. Conclusion: In this study, we found that NLRP3 could promote the migration and invasion of colorectal cancer cells in the inflammatory microenvironment. Galloflavin could inhibit the malignant behavior of colorectal cancer cells by targeting NLRP3.

17.
Front Pharmacol ; 12: 802304, 2021.
Article in English | MEDLINE | ID: mdl-34938199

ABSTRACT

[This corrects the article DOI: 10.3389/fphar.2020.570776.].

18.
Front Pharmacol ; 11: 570776, 2020.
Article in English | MEDLINE | ID: mdl-33192512

ABSTRACT

OBJECTIVE: In this study, we mainly explored the mechanism and target of the anti-inflammatory effects of Aureusidin (Aur) in acute liver injury. METHODS: Lipopolysaccharide (LPS) was used to induce inflammatory injury in Kupffer cells (KCs) in vitro. After Aur treatment with gradient concentration, flow cytometry, propidium iodide (PI) staining, and Hoechst 33342 staining were used to detect the apoptotic level of KCs, and an enzyme-linked immunosorbent assay (ELISA) was used to detect the expression levels of inflammatory factors, including Interleukin-1ß (IL-1ß), Interleukin-18 (IL-18), and tumor necrosis factor alpha (TNF-α). Western blot was used to detect the expression of toll-like receptor 4 (TLR4), myeloid differentiation protein-2 (MD2), MyD88, and p-P65. Aur was labeled with biotin, followed by a pull-down assay to detect its binding with MD2. Moreover, D-GalN/LPS was used to induce acute liver injury in mice in vitro, followed by Aur treatment by gavage. H&E staining was used to detect the pathological changes of liver tissue, an IF assay was used to detect the expression of MD2, Western blot was used to detect the expression of relevant proteins. RESULTS: Aur pretreatment could significantly inhibit LPS-induced KC injury, downregulate the apoptotic level, inhibit the expression of inflammatory factors, decrease the level of MDA, and downregulate the expression of MD2 in cells. Aur could inhibit the activation level of TLR4/MD2-NF-κB in a dose-dependent pattern, a high dose of Aur had a superior effect compared to low-dose Aur. In the case of MD2 deletion, the effects of Aur were suppressed. Additionally, pull-down and co-immunoprecipitation assays show that Aur can bind with the MD2 protein to inhibit the activation of TLR4/MD2-NF-κB. Results of mice experiments also showed that Aur could relieve liver injury, decrease the levels of ALT and AST, and simultaneously downregulate the levels of inflammatory factors in tissues and peripheral blood. CONCLUSION: We found that Aur exerted an anti-inflammatory effect by directly targeting the MD2 protein, further inhibiting the expression of TLR4/MD2-NF-κB, thereby relieving acute liver injury. Therefore, Aur might be a potential inhibitor for MD2.

SELECTION OF CITATIONS
SEARCH DETAIL